Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Bioorg Chem ; 119: 105522, 2022 02.
Article in English | MEDLINE | ID: mdl-34864279

ABSTRACT

Six new lignans with various type of linkage between two C6-C3 fragments (1a, 1b, 2a, 2b, 3, 4), two new meroterpenoids (5, 6) and 24 known compounds (7-30) were isolated from an EtOH extract of the stems and leaves of Piper puberulum. The absolute configurations of enantiomers 1a and 1b were determined by single-crystal X-ray diffraction analysis, 2a and 2b were determined by comparing their calculated and experimental ECD spectra. Biogenetically, all the new lignans may come from the polymerization of two molecules of hydroxychavicol (30). In the anti-neuroinflammation activity assay, the IC50 values of fifteen compounds were lower than those of the positive control minocycline, and compound 1a showed good activity, but its enantiomer 1b showed no activity. Compound 1a have notable anti-neuroinflammatory activity, and can significantly decrease mRNA levels of proinflammatory cytokines (IL-1ß, IL-6, TNF-α) in a dose-dependent manner.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Lignans/pharmacology , Nitric Oxide/antagonists & inhibitors , Piper/chemistry , Plant Extracts/pharmacology , Terpenes/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/isolation & purification , Cell Line , Crystallography, X-Ray , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Dose-Response Relationship, Drug , Lignans/chemistry , Lignans/isolation & purification , Mice , Models, Molecular , Molecular Structure , Nitric Oxide/metabolism , Plant Extracts/chemistry , Plant Extracts/isolation & purification , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , Stereoisomerism , Structure-Activity Relationship , Terpenes/chemistry , Terpenes/isolation & purification
2.
EBioMedicine ; 74: 103691, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34773891

ABSTRACT

Alzheimer's disease (AD) represents a particular therapeutic challenge because its aetiology is very complex, with dynamic progression from preclinical to clinical stages. Several potential therapeutic targets and strategies were tested for AD, in over 2000 clinical trials, but no disease-modifying therapy exists. This failure indicates that AD, as a multifactorial disease, may require multi-targeted approaches and the delivery of therapeutic molecules to the right place and at the right disease stage. Opportunities to meet the challenges of AD therapy appear to come from recent progress in knowledge and methodological advances in the design, synthesis, and targeting of brain mRNA and microRNA with synthetic antisense oligonucleotides (ASOs). Several types of ASOs allow the utilisation of different mechanisms of posttranscriptional regulation and offer enhanced effects over alternative therapeutics. This article reviews ASO-based approaches and targets in preclinical and clinical trials for AD, and presents the future perspective on ASO therapies for AD.


Subject(s)
Alzheimer Disease/genetics , MicroRNAs/genetics , Oligonucleotides, Antisense/pharmacology , RNA, Messenger/genetics , Alzheimer Disease/drug therapy , Brain Chemistry , Gene Expression Regulation/drug effects , Gene Regulatory Networks/drug effects , Humans , MicroRNAs/antagonists & inhibitors , Molecular Targeted Therapy , RNA, Messenger/antagonists & inhibitors
3.
Bioorg Med Chem Lett ; 41: 127986, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33766770

ABSTRACT

Monoacylglycerol lipase (MAGL) is the major enzyme that catalyzes the hydrolysis of monoacylglycerols (MAGs). MAGL is responsible for degrading 2-arachidonoylglycerol (2-AG) to arachidonic acid (AA) and glycerol in the brain and specific tissues. The inhibition of MAGL could attenuate the inflammatory response. Here, we report a series of reversible non-covalent MAGL inhibitors via virtual screening combined with biochemical analysis. The hit, DC630-8 showed low-micromolar activity against MAGL in vitro, and exhibited significant anti-inflammatory effects.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cytokines/antagonists & inhibitors , Drug Discovery , Enzyme Inhibitors/pharmacology , Molecular Docking Simulation , Monoacylglycerol Lipases/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/chemistry , Cytokines/biosynthesis , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Mice , Molecular Structure , Monoacylglycerol Lipases/metabolism , RAW 264.7 Cells , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , Structure-Activity Relationship
4.
Molecules ; 26(4)2021 Feb 20.
Article in English | MEDLINE | ID: mdl-33672773

ABSTRACT

A caloric surplus and a sedentary lifestyle are undoubtedly known to be the leading causes of obesity. Natural products represent valuable allies to face this problematic issue. This study was planned to assess the effect of a white grape (Vitis vinifera) juice extract (WGJe) in diet-induced obese zebrafish (Danio rerio). Fish were divided into four different diet groups: (i) normally fed (NF); (ii) overfed (OF); (iii) WGJe-supplemented NF (5 mL/L in fish water); (iv) WGJe-supplemented OF. Body mass index (BMI) was extrapolated each week. After the fourth week, euthanized zebrafish were processed for both microscopic evaluations and gene expression analyses. OF zebrafish showed higher BMI values with respect to NF counterparts, an effect that was hindered by WGJe treatment. Moreover, histological analyses showed that the area of the adipose tissue, as well as the number, size, and density of adipocytes was significantly higher in OF fish. On the other hand, WGJe was able to avoid these outcomes both at the subcutaneous and visceral levels, albeit to different extents. At the gene level, WGJe restored the altered levels of ghrelin and leptin of OF fish both in gut and brain. Overall, our results support the anti-obesity property of WGJe, suggesting its potential role in weight management.


Subject(s)
Adipocytes/drug effects , Fats/antagonists & inhibitors , Ghrelin/antagonists & inhibitors , Leptin/antagonists & inhibitors , Plant Extracts/pharmacology , Vitis/chemistry , Animals , Disease Models, Animal , Fats/metabolism , Fruit and Vegetable Juices/analysis , Ghrelin/genetics , Ghrelin/metabolism , Leptin/genetics , Leptin/metabolism , Molecular Structure , Obesity/drug therapy , Obesity/metabolism , Plant Extracts/administration & dosage , Plant Extracts/chemistry , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Zebrafish
5.
Molecules ; 26(5)2021 Feb 25.
Article in English | MEDLINE | ID: mdl-33669031

ABSTRACT

Response surface methodology was employed to optimize the ultrasound-assisted extraction (UAE) conditions for simultaneous optimization of dependent variables, including DPPH radical scavenging activity (RSA), tyrosinase activity inhibition (TAI), and collagenase activity inhibition (CAI) of peanut shell extracts. The effects of the main variables including extraction time (5.0~55.0 min, X1), extraction temperature (26.0~94.0 °C, X2), and ethanol concentration (0.0%~99.5%, X3) were optimized. Based on experimental values from each condition, quadratic regression models were derived for the prediction of optimum conditions. The coefficient of determination (R2) of the independent variable was in the range of 0.89~0.96, which demonstrates that the regression model is suitable for the prediction. In predicting optimal UAE conditions based on the superimposing method, extraction time of 31.2 min, extraction temperature of 36.6 °C, and ethanol concentration of 93.2% were identified. Under these conditions, RSA of 74.9%, TAI of 50.6%, and CAI of 86.8% were predicted, showing good agreement with the experimental values. A reverse transcription polymerase chain reaction showed that peanut shell extract decreased mRNA levels of tyrosinase-related protein-1 and matrix metalloproteinase-3 genes in B16-F0 cell. Therefore, we identified the skin-whitening and anti-wrinkle effects of peanut shell extracts at protein as well as gene expression levels, and the results show that peanut shell is an effective cosmetic material for skin-whitening and anti-wrinkle effects. Based on this study, peanut shell, which was considered a byproduct, can be used for the development of healthy foods, medicines, and cosmetics.


Subject(s)
Antioxidants/pharmacology , Arachis/chemistry , Plant Extracts/pharmacology , Skin Lightening Preparations/pharmacology , Ultrasonic Waves , Animals , Antioxidants/chemistry , Antioxidants/isolation & purification , Biphenyl Compounds/antagonists & inhibitors , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/genetics , Oxidoreductases/metabolism , Picrates/antagonists & inhibitors , Plant Extracts/chemistry , Plant Extracts/isolation & purification , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Skin Aging/drug effects , Skin Lightening Preparations/chemistry , Skin Lightening Preparations/isolation & purification , Tumor Cells, Cultured
6.
Nucleic Acid Ther ; 30(4): 198-206, 2020 08.
Article in English | MEDLINE | ID: mdl-32589506

ABSTRACT

The aim of this study was to assess the effect of volanesorsen on the corrected QT (QTc) interval. This thorough QT study enrolled 52 healthy male and female subjects who were randomized at a single site in a four-way crossover study. Subjects were randomly assigned to 1 of 12 treatment sequences and crossed over into four treatment periods over the course of which each subject was to receive a single therapeutic dose of volanesorsen as a 300 mg subcutaneous (SC) injection, a single supratherapeutic dose of volanesorsen as 300 mg intravenous (IV) infusion, a single oral (PO) dose of moxifloxacin (positive control), and placebo dose. The study demonstrated that volanesorsen 300 mg SC and 300 mg IV did not have a clinically relevant effect on ΔΔQTcF exceeding 10 ms. The largest mean effect at any postdose time point was 3.0 ms (90% confidence interval [CI]: 0.8-5.2) after SC dosing and 1.8 ms (90% CI -0.4 to 4.0) after IV dosing. Volanesorsen, at the studied therapeutic and supratherapeutic doses, does not have a clinically meaningful effect on the QTc.


Subject(s)
Apolipoprotein C-III/genetics , Hypertriglyceridemia/therapy , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides/administration & dosage , Adult , Apolipoprotein C-III/antagonists & inhibitors , Dose-Response Relationship, Drug , Double-Blind Method , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/pathology , Electrocardiography , Female , Healthy Volunteers , Humans , Hypertriglyceridemia/diagnostic imaging , Hypertriglyceridemia/genetics , Hypertriglyceridemia/pathology , Male , Moxifloxacin/administration & dosage , Oligonucleotides/adverse effects , Oligonucleotides, Antisense/adverse effects , Placebo Effect , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , Triglycerides/antagonists & inhibitors , Triglycerides/blood
7.
J Biochem Mol Toxicol ; 33(12): e22406, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31593353

ABSTRACT

Soybean Bowman-Birk protease inhibitor (BBI) and genistein, two biological compounds from soybean, are well-known for their anti-inflammatory, antioxidant, and anticancer activities. The aim of this study was designing a BBI-genistein conjugate and then investigating its protective effect on lipopolysaccharide (LPS)-induced inflammation in BALB/c mice, compared with the effects of combination of BBI and genistein. BBI was purified from soybean and the BBI-genistein conjugate was synthesized. The BALB/c mice were intraperitoneally treated 2 hours before LPS induction. Our results showed that treatment with the combination of BBI and genistein greatly led to more reduced serum levels of tumor necrosis factor (TNF)-α and interferon (IFN)-γ compared with the treatments of BBI alone, the BBI-genistein conjugate, and genistein alone, respectively. Moreover, the expression of TNF-α and IFN-γ in the splenocytes was significantly downregulated along with improving host survival against the LPS-induced lethal endotoxemia in the same way. Our data support a new combined therapy using BBI and genistein, as natural anti-inflammatory agents, to develop a new drug for inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Endotoxemia/drug therapy , Genistein/therapeutic use , Glycine max/chemistry , Plant Extracts/therapeutic use , Trypsin Inhibitor, Bowman-Birk Soybean/therapeutic use , Animals , Drug Combinations , Endotoxemia/chemically induced , Genistein/administration & dosage , Inflammation/metabolism , Injections, Intraperitoneal , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/blood , Kaplan-Meier Estimate , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred BALB C , RNA, Messenger/antagonists & inhibitors , Spleen/pathology , Survival Rate , Treatment Outcome , Trypsin Inhibitor, Bowman-Birk Soybean/administration & dosage , Trypsin Inhibitor, Bowman-Birk Soybean/isolation & purification , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/blood
8.
Drug Des Devel Ther ; 13: 975-990, 2019.
Article in English | MEDLINE | ID: mdl-30988600

ABSTRACT

BACKGROUND: Emodin has recently been reported to have a powerful antiinflammatory effect, protecting the myocardium against ischemia/reperfusion (I/R) injury. Pyroptosis is a proinflammatory programmed cell death that is related to many diseases. The present study investigated the effect of emodin on pyroptosis in cardiomyocytes. MATERIALS AND METHODS: Sprague Dawley rats were randomly divided into sham, I/R, and I/R+Emodin groups. I/R model was subjected to 30 minutes' ligation of left anterior descending coronary artery, followed by 2 hours of reperfusion. Cardiomyocytes were exposed to hypoxic conditions for 1 hour and normoxic conditions for 2 hours. The level of the pyroptosis was detected by Western blot, real-time PCR analysis, and ELISA. RESULTS: The level of gasdermin D-N domains was upregulated in cardiomyocytes during I/R or hypoxia/reoxygenation (H/R) treatment. Moreover, emodin increased the rate of cell survival in vitro and decreased the myocardial infarct size in vivo via suppressing the levels of I/R-induced pyroptosis. Additionally, the expression of TLR4, MyD88, phospho-IκBα, phospho-NF-κB, and the NLRP3 inflammasome was significantly upregulated in cardiomyocytes subjected to H/R treatment, while emodin suppressed the expression of these proteins. CONCLUSION: This study confirms that emodin treatment was able to alleviate myocardial I/R injury and inhibit pyroptosis in vivo and in vitro. The inhibitory effect of emodin on pyroptosis was mediated by suppressing the TLR4/MyD88/NF-κB/NLRP3 inflammasome pathway. Therefore, emodin may provide an alternative treatment for myocardial I/R injury.


Subject(s)
Apoptosis Regulatory Proteins/antagonists & inhibitors , Emodin/pharmacology , Myocardial Reperfusion Injury/drug therapy , Myocytes, Cardiac/drug effects , Pyroptosis/drug effects , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Survival/drug effects , Dose-Response Relationship, Drug , Emodin/chemistry , Intracellular Signaling Peptides and Proteins , Male , Medicine, Chinese Traditional , Molecular Structure , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phosphate-Binding Proteins , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Rheum/chemistry , Structure-Activity Relationship
9.
Biosci Biotechnol Biochem ; 83(7): 1181-1192, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31032736

ABSTRACT

Interaction between foods and drugs is an important consideration in pharmaceutical therapy. Therefore, here, we examined the suppressive effects of the extracts from seven edible herbs on the induction of CYP3A4 gene expression in rifampicin-treated HepG2 cells. We evaluated the structure and suppressive activity of the most effective active compound isolated from dried peppermint (Mentha piperita L.). The structure of the compound was identified as that of pheophorbide a based on spectroscopic data. It suppressed the induction of CYP3A4 mRNA expression by rifampicin in a dose-dependent manner. Quantitative high-performance liquid chromatography showed that 2 g of dry leaves 0.43 mg in one cup of peppermint tea. These findings demonstrate that pheophorbide a suppresses the induction of CYP3A4 mRNA expression in rifampicin-treated HepG2 cells. Pheophorbide is known to cause photosensitivity. However, the effective dose of pheophorbide a that had a suppressive effect was very low, indicating a high safety margin. Abbreviations: DAD: diode array detector; DMEM: Dulbecco's modified Eagle's medium; ELISA: enzyme-linked immunosorbent assay; HPLC: high-performance liquid chromatography; PCR: polymerase chain reaction; PXR: pregnane X receptor; CAR: constitutive androstane receptor; AHR: aryl hydrocarbon receptor; TLC: thin-layer chromatography.


Subject(s)
Antibiotics, Antitubercular/pharmacology , Chlorophyll/analogs & derivatives , Cytochrome P-450 CYP3A/genetics , Mentha piperita/chemistry , Plant Extracts/pharmacology , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , Rifampin/pharmacology , Cell Survival/drug effects , Chlorophyll/chemistry , Chlorophyll/pharmacology , Chromatography, High Pressure Liquid , Hep G2 Cells , Humans , Molecular Structure , Plant Extracts/chemistry , Proton Magnetic Resonance Spectroscopy , Structure-Activity Relationship
10.
Clin Transl Sci ; 12(2): 98-112, 2019 03.
Article in English | MEDLINE | ID: mdl-30706991

ABSTRACT

The therapeutic pathways that modulate transcription mechanisms currently include gene knockdown and splicing modulation. However, additional mechanisms may come into play as more understanding of molecular biology and disease etiology emerge. Building on advances in chemistry and delivery technology, oligonucleotide therapeutics is emerging as an established, validated class of drugs that can modulate a multitude of genetic targets. These targets include over 10,000 proteins in the human genome that have hitherto been considered undruggable by small molecules and protein therapeutics. The approval of five oligonucleotides within the last 2 years elicited unprecedented excitement in the field. However, there are remaining challenges to overcome and significant room for future innovation to fully realize the potential of oligonucleotide therapeutics. In this review, we focus on the translational strategies encompassing preclinical evaluation and clinical development in the context of approved oligonucleotide therapeutics. Translational approaches with respect to pharmacology, pharmacokinetics, cardiac safety evaluation, and dose selection that are specific to this class of drugs are reviewed with examples. The mechanism of action, chemical evolution, and intracellular delivery of oligonucleotide therapies are only briefly reviewed to provide a general background for this class of drugs.


Subject(s)
Genetic Therapy/methods , RNA, Messenger/genetics , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacokinetics , Clinical Trials as Topic , Drug Approval , Drug Delivery Systems/methods , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Humans , Oligoribonucleotides, Antisense/administration & dosage , Oligoribonucleotides, Antisense/genetics , Oligoribonucleotides, Antisense/pharmacokinetics , RNA Interference , RNA Stability/drug effects , RNA, Messenger/agonists , RNA, Messenger/antagonists & inhibitors , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacokinetics , Transcription, Genetic/drug effects
11.
Mol Pharm ; 15(12): 5781-5792, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30392378

ABSTRACT

Polyglutamine diseases are a set of progressive neurodegenerative disorders caused by misfolding and aggregation of mutant CAG RNA and polyglutamin protein. To date, there is a lack of effective therapeutics that can counteract the polyglutamine neurotoxicity. Two peptidylic inhibitors, QBP1 and P3, targeting the protein and RNA toxicities, respectively, have been previously demonstrated by us with combinational therapeutic effects on the Drosophila polyglutamine disease model. However, their therapeutic efficacy has never been investigated in vivo in mammals. The current study aims to (a) develop a brain-targeting delivery system for both QBP1 and L1P3V8 (a lipidated variant of P3 with improved stability) and (b) evaluate their therapeutic effects on the R6/2 transgenic mouse model of polyglutamine disease. Compared with intravenous administration, intranasal administration of QBP1 significantly increased its brain-to-plasma ratio. In addition, employment of a chitosan-containing in situ gel for the intranasal administration of QBP1 notably improved its brain concentration for up to 10-fold. Further study on intranasal cotreatment with the optimized formulation of QBP1 and L1P3V8 in mice found no interference on the brain uptake of each other. Subsequent efficacy evaluation of 4-week daily QBP1 (16 µmol/kg) and L1P3V8 (6 µmol/kg) intranasal cotreatment in the R6/2 mice demonstrated a significant improvement on the motor coordination and explorative behavior of the disease mice, together with a full suppression on the RNA- and protein-toxicity markers in their brains. In summary, the current study developed an efficient intranasal cotreatment of the two peptidylic inhibitors, QBP1 and L1P3V8, for their brain-targeting, and such a novel therapeutic strategy was found to be effective on a transgenic polyglutamine disease mouse model.


Subject(s)
Carrier Proteins/administration & dosage , Heredodegenerative Disorders, Nervous System/drug therapy , Oligopeptides/administration & dosage , Peptides/administration & dosage , Peptides/metabolism , RNA, Messenger/antagonists & inhibitors , Administration, Intranasal , Animals , Behavior, Animal/drug effects , Brain/metabolism , Carrier Proteins/pharmacokinetics , Disease Models, Animal , Drug Administration Schedule , Drug Evaluation, Preclinical , Drug Therapy, Combination/methods , Heredodegenerative Disorders, Nervous System/genetics , Heredodegenerative Disorders, Nervous System/pathology , Humans , Intercellular Signaling Peptides and Proteins , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligopeptides/pharmacokinetics , Peptides/pharmacokinetics , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Trinucleotide Repeat Expansion/genetics
12.
Int J Mol Med ; 41(4): 2128-2138, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29393411

ABSTRACT

Human apurinic/apyrimidinic endonuclease 1 (APE1) is a ubiquitous multifunctional protein, which possesses DNA repair and redox activities. High levels of APE1 are associated with chemo­ and radioresistance, and poor prognosis in various types of cancer, including non­small cell lung cancer (NSCLC). Bu­Fei decoction (BFD) is a traditional Chinese herbal formula, which is believed to supplement Qi, clear away heat and nourish the lungs. BFD and modified Bu­Fei decoction (MBFD) have been used in China to treat patients with lung cancer. The present study aimed to evaluate the potential antitumor effects of BFD and MBFD on NSCLC in vitro and in vivo. In addition, the possible contribution of APE1 was examined. MTT assay was used to investigated the anti-tumor activity of BFD and MBFD on H1975 and H292 NSCLC cell lines. The DNA damage of cells in the control and the experimental groups was detected using comet assay. The in vivo anti-tumor effects of BFD and MBFD were evaluated in a NSCLC tumor nude mouse xenograft model. Polymerase chain reaction (PCR), reverse transcription­quantitative PCR (RT­qPCR) analysis and western blot analysis were applied to analyze the mRNA and protein expression levels of APE1 in H1975 and H292 cells, so as to the xenograft tumor tissues. The concentration of APE1 in mice plasma was determined using enzyme linked immunosorbent assay (ELISA). In vitro, BFD and MBFD inhibited the growth of cultured H1975 and H292 NSCLC cells. The results of a comet assay revealed that BFD and MBFD increased DNA damage. Furthermore, the expression levels of APE1 were decreased in response to BFD and MBFD at the mRNA and protein levels. In mice carrying NSCLC xenografts, BFD and MBFD inhibited tumor growth and decreased APE1 expression. In addition, in normal human lung bronchial epithelial BEAS­2B cells, the half maximal inhibitory concentrations of BFD and MBFD were much higher compared with in NSCLC cells, and they had no effect on DNA damage. These results suggested that BFD and MBFD may inhibit the growth of NSCLC, possibly by inhibiting APE1 expression.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Proliferation/drug effects , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , Down-Regulation/drug effects , Drugs, Chinese Herbal/therapeutic use , Lung Neoplasms/drug therapy , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle/drug effects , Cell Line , Cell Line, Tumor , DNA Repair/drug effects , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Drugs, Chinese Herbal/pharmacology , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice, Inbred BALB C , Mice, Nude , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics
13.
Biomed Pharmacother ; 97: 1-8, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29080449

ABSTRACT

Peumus boldus (P. boldus) is a medicinal plant popularly used in the treatment of gastrointestinal disorders. P. boldus aqueous extract is rich in phenolic compounds and alkaloids that possess antiinflammatory and antioxidant effects. In the present study, the potential protective effect of P. boldus against Cu2+-induced toxicity was investigated. Adult Drosophila melanogaster were exposed to Cu2+ (1mM and 3mM) and/or P. boldus aqueous extract (5mg/mL) in the food during 4days. Cu2+-fed flies had impairment in the negative geotaxis performance (i.e. motor climbing capability) as well as a higher incidence of mortality when compared to the control group. P. boldus co-treatment afforded protection against the Cu2+-induced toxicity. Acetylcholinesterase (AChE) and glutathione S-transferase (GST) activity decreased significantly in D. melanogaster after Cu2+ exposure. P. boldus co-exposure for 4days restored enzyme activities to control levels. In addition, Cu2+ exposure caused a significant increase in the mRNA levels of antioxidant enzymes, superoxide dismutase (Sod1), catalase (Cat), thioredoxin reductase (TrxR1) and nuclear factor erythroid 2-related factor 2 (Nrf2), as well as increased the mRNA levels of acetylcholinesterase (Ace). The expression of P-type ATPase (Atp7A) and copper uptake protein 1 (Ctr1A) mRNAs were up-regulated in D. melanogaster exposed to Cu2+. The co-treatment with P. boldus blunted Cu2+-induced up-regulation of Atp7A and down-regulated Ctr1A mRNA expression. These findings suggest that P. boldus extracts reduce Cu2+-induced toxicity but not Cu2+ absorption in D. melanogaster. Consequently, P. boldus can be a potential therapeutical alternative for modulating Cu2+-associated toxicity.


Subject(s)
Copper/toxicity , Oxidative Stress/drug effects , Peumus , Plant Extracts/pharmacology , Animals , Copper/metabolism , Drosophila melanogaster , Oxidative Stress/physiology , Plant Extracts/isolation & purification , Plant Leaves , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , Survival Rate/trends
14.
Int J Mol Sci ; 19(1)2017 Dec 28.
Article in English | MEDLINE | ID: mdl-29283381

ABSTRACT

A growing body of studies has documented the pathological influence of impaired alternative splicing (AS) events on numerous diseases, including cancer. In addition, the generation of alternatively spliced isoforms is frequently noted to result in drug resistance in many cancer therapies. To gain comprehensive insights into the impacts of AS events on cancer biology and therapeutic developments, this paper highlights recent findings regarding the therapeutic routes of targeting alternative-spliced isoforms and splicing regulators to treatment strategies for distinct cancers.


Subject(s)
Alternative Splicing/drug effects , Antineoplastic Agents/therapeutic use , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/therapy , RNA Splicing Factors/antagonists & inhibitors , RNA, Messenger/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinogenesis/pathology , Caspase 9/genetics , Caspase 9/metabolism , Cyclin D1/antagonists & inhibitors , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclohexylamines/therapeutic use , Epoxy Compounds/therapeutic use , Humans , Macrolides/therapeutic use , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oligonucleotides/genetics , Oligonucleotides/metabolism , Oligonucleotides/therapeutic use , Pyrans/therapeutic use , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Spiro Compounds/therapeutic use , Spliceosomes/drug effects , Spliceosomes/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
15.
Histochem Cell Biol ; 147(1): 63-73, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27600718

ABSTRACT

We aimed to study the impact of altered thyroid status on myocardial expression of electrical coupling protein connexin-43 (Cx43), the susceptibility of rats to ventricular fibrillation (VF) and the effects of antioxidant-rich red palm oil (RPO). Adult male and female euthyroid, hyperthyroid (treated with T3/T4), hypothyroid (treated with methimazole) Wistar rats supplemented or not with RPO for 6 weeks were used. Function of isolated perfused heart and VF threshold were determined. Left ventricular tissue was used for assessment of mRNA and protein levels of Cx43, its phosphorylated forms and topology. Protein kinase C signaling (PKC) and gene transcripts of some proteins related to cardiac arrhythmias were assessed. Hyperthyroid state resulted in decrease of total and phosphorylated forms of Cx43 and suppression of PKC-ε expression in males and females, decrease of Cx43 mRNA in females, decrease of VF threshold and increase of functional parameters in male rat hearts. In contrast, hypothyroid status resulted in the increase of total and phosphorylated forms of Cx43, enhancement PKC-ε expression in males and females, increase of Cx43 mRNA in females, increase of VF threshold and decrease of functional parameters in male rat hearts. Function of the heart was partially normalized by RPO intake, which also enhanced myocardial Cx43 and PKC-ε expression as well as increased VF threshold in hyperthyroid male rats. We conclude that there is an inverse relationship between myocardial expression of Cx43, including its functional phosphorylated forms, and susceptibility of male rat hearts to VF in condition of altered thyroid status. RPO intake partly ameliorated adverse changes caused by excess of thyroid hormones.


Subject(s)
Arrhythmias, Cardiac/drug therapy , Connexin 43/genetics , Heart/drug effects , Myocardium/metabolism , Plant Oils/pharmacology , Thyroid Gland/drug effects , Administration, Oral , Animals , Arrhythmias, Cardiac/metabolism , Connexin 43/antagonists & inhibitors , Connexin 43/metabolism , Female , Male , Palm Oil , Plant Oils/administration & dosage , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Thyroid Gland/metabolism
16.
Amyloid ; 23(2): 109-18, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27033334

ABSTRACT

ATTR amyloidosis is a systemic, debilitating and fatal disease caused by transthyretin (TTR) amyloid accumulation. RNA interference (RNAi) is a clinically validated technology that may be a promising approach to the treatment of ATTR amyloidosis. The vast majority of TTR, the soluble precursor of TTR amyloid, is expressed and synthesized in the liver. RNAi technology enables robust hepatic gene silencing, the goal of which would be to reduce systemic levels of TTR and mitigate many of the clinical manifestations of ATTR that arise from hepatic TTR expression. To test this hypothesis, TTR-targeting siRNAs were evaluated in a murine model of hereditary ATTR amyloidosis. RNAi-mediated silencing of hepatic TTR expression inhibited TTR deposition and facilitated regression of existing TTR deposits in pathologically relevant tissues. Further, the extent of deposit regression correlated with the level of RNAi-mediated knockdown. In comparison to the TTR stabilizer, tafamidis, RNAi-mediated TTR knockdown led to greater regression of TTR deposits across a broader range of affected tissues. Together, the data presented herein support the therapeutic hypothesis behind TTR lowering and highlight the potential of RNAi in the treatment of patients afflicted with ATTR amyloidosis.


Subject(s)
Amyloid Neuropathies, Familial/therapy , Liver/metabolism , Prealbumin/antagonists & inhibitors , RNA, Messenger/antagonists & inhibitors , RNA, Small Interfering/administration & dosage , Amyloid Neuropathies, Familial/genetics , Amyloid Neuropathies, Familial/metabolism , Amyloid Neuropathies, Familial/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Benzoxazoles/pharmacology , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Gene Expression , Humans , Liver/pathology , Macaca fascicularis , Male , Mice , Mice, Transgenic , Prealbumin/genetics , Prealbumin/metabolism , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacokinetics
17.
Biochemistry ; 55(13): 1977-88, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-26959335

ABSTRACT

We have examined the abilities of three complementary γ-peptide nucleic acid (γPNA) oligomers to invade an RNA G-quadruplex and potently inhibit translation of a luciferase reporter transcript containing the quadruplex-forming sequence (QFS) within its 5'-untranslated region. All three γPNA oligomers bind with low nanomolar affinities to an RNA oligonucleotide containing the QFS. However, while all probes inhibit translation with low to midnanomolar IC50 values, the γPNA designed to hybridize to the first two G-tracts of the QFS and adjacent 5'-overhanging nucleotides was 5-6 times more potent than probes directed to either the 3'-end or internal regions of the target at 37 °C. This position-dependent effect was eliminated after the probes and target were preincubated at an elevated temperature prior to translation, demonstrating that kinetic effects exert significant control over quadruplex invasion and translation inhibition. We also found that antisense γPNAs exhibited similarly potent effects against luciferase reporter transcripts bearing QFS motifs having G2, G3, or G4 tracts. Finally, our results indicate that γPNA oligomers exhibit selectivity and/or potency higher than those of other antisense molecules such as standard PNA and 2'-OMe RNA previously reported to target G-quadruplexes in RNA.


Subject(s)
Drug Design , G-Quadruplexes/drug effects , Oligonucleotides, Antisense/pharmacology , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/antagonists & inhibitors , 5' Untranslated Regions/drug effects , Amino Acid Motifs , Animals , GTP Phosphohydrolases/genetics , Genes, Reporter/drug effects , Glycine/analogs & derivatives , Glycine/chemistry , Humans , Kinetics , Membrane Proteins/genetics , Nucleic Acid Conformation , Nucleic Acid Denaturation , Protein Synthesis Inhibitors/chemistry , Protein Synthesis Inhibitors/metabolism , RNA Stability/drug effects , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Rabbits , Reticulocytes/enzymology , Reticulocytes/metabolism
18.
Neoplasma ; 63(3): 351-61, 2016.
Article in English | MEDLINE | ID: mdl-26925795

ABSTRACT

Epithelial-mesenchymal transition (EMT) is considered as the most important mechanism that underlies the initiation of cancer metastasis. Here we report that Physicon 8-O-ß-glucopyranoside (PG), a major active ingredient from a traditional Chinese herbal medicine Rumex japonicus Houtt, is capable of preventing human colorectal cancer cells from hypoxia-induced EMT. The treatment of the cells with PG reversed the EMT-related phenotype that has the morphological changes, down-regulation of E-cadherin, and hypoxia-induced cell migration and invasion. The effect was mediated at least in part by inhibiting the mRNA and protein expressions of EMMPRIN via modulation of PTEN/Akt/HIF-1α pathway. In addition, we found that PG-mediated prevention of EMT involved blockade of the hypoxia-induced up-regulation of Snail, Slug and Twist. In summary, this study showed that PG can prevent EMT induced by hypoxia, the environment that commonly exists in the center of a solid tumor. Given the low toxicity of PG to the healthy tissues, our study suggests that PG can serve as a safe therapeutic agent for suppressing cancer metastasis.


Subject(s)
Basigin/antagonists & inhibitors , Cell Hypoxia/drug effects , Colorectal Neoplasms/drug therapy , Emodin/analogs & derivatives , Epithelial-Mesenchymal Transition/drug effects , Glucosides/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Basigin/biosynthesis , Basigin/genetics , Cell Movement/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Emodin/pharmacology , HCT116 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Signal Transduction/drug effects
19.
Cell Mol Biol (Noisy-le-grand) ; 61(8): 37-43, 2015 Dec 14.
Article in English | MEDLINE | ID: mdl-26667771

ABSTRACT

Berberine is an isoquinoline alkaloid found in several plant species like famous chinese herb, Rhizoma coptidis which has been used locally as a strong gastrointestinal remedy for thousands of years. The inhibitory effects of berberine on tumor progression properties have been reported before. In this study, we investigated the effect of berberine on an esophageal cancer cell line, KYSE-30 with emphasis on its effects on the expression of certain chemokine receptors. The cytotoxic effect of berberine on KYSE-30 cells was analyzed by MTT assay. In vitro cell migration assay was also applied to the treated cells and the expression levels of the selected chemokine receptors (CXCR4 and CCR7) was measured at mRNA level. A retarded growth, associated with increasing concentrations of berberine, was obvious. On the other hand, the migration rate of the cells was decreased when they were treated with different concentrations of berberine and the expression levels of the two chemokine receptors, involved in the migration and metastasis of esophageal cancer cells, were decreased following the same treatments. With these results, we tend to conclude that berberine might be a proper candidate for further investigations, by targeting the chemokine receptors, and possible applications as anti-metastatic agent in cancer studies.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Berberine/pharmacology , Biomarkers, Tumor/antagonists & inhibitors , Drugs, Chinese Herbal/pharmacology , Gene Expression Regulation, Neoplastic , RNA, Messenger/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/isolation & purification , Berberine/isolation & purification , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/isolation & purification , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Esophagus/drug effects , Esophagus/metabolism , Esophagus/pathology , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, CCR7/antagonists & inhibitors , Receptors, CCR7/genetics , Receptors, CCR7/metabolism , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Signal Transduction
20.
Mol Med Rep ; 12(4): 5580-6, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26166377

ABSTRACT

Berberine, which is a well­known drug used in traditional medicine, has been demonstrated to exert diverse pharmacological effects, including anti­inflammatory effects. However, whether berberine can affect the production of inflammatory molecules in vascular endothelial cells remains to be elucidated. Therefore, the present study aimed to determine the effects of berberine, and the underlying molecular mechanisms of these effects. The effect of berberine on tumor necrosis factor (TNF)­α­induced inflammatory molecule expression was examined in cultured human aortic endothelial cells (HAECs). The HAECs were stimulated with TNF­α and incubated with or without berberine. The activation of nuclear factor (NF)­κB and adenosine monophosphate­activated protein kinase (AMPK) were analyzed using western blotting, and the protein secretion of intercellular adhesion molecule (ICAM)­1 and monocyte chemoattractant protein (MCP)­1 was measured using ELISA kits. The mRNA expression levels of ICAM­1 and MCP­1 were analyzed using reverse transcription­quantitative polymerase chain reaction. The results of the present study demonstrated that berberine significantly inhibited the TNF­α­induced expression of ICAM­1 and MCP­1, as well as the activation of NF­κB in the HAECs. These effects were attenuated following co­treatment with AMPK inhibitor compound C, or specific small interfering RNAs. In conclusion, the results of the present study indicated that berberine inhibits the TNF­α­induced expression of ICAM­1 and MCP­1, and the activation of NF­κB in HAECs in vitro, possibly through the AMPK­dependent pathway.


Subject(s)
AMP-Activated Protein Kinases/genetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Berberine/pharmacology , Endothelial Cells/drug effects , NF-kappa B/genetics , Tumor Necrosis Factor-alpha/pharmacology , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Cell Line , Chemokine CCL2/agonists , Chemokine CCL2/antagonists & inhibitors , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression Regulation , Humans , Inflammation , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , NF-kappa B/agonists , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/agonists , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL